RhoA, Rac1, and Cdc42 differentially regulate αSMA and collagen I expression in mesenchymal stem cells

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Standard

RhoA, Rac1, and Cdc42 differentially regulate αSMA and collagen I expression in mesenchymal stem cells. / Ge, Jianfeng; Burnier, Laurent; Adamopoulou, Maria; Kwa, Mei Qi; Schaks, Matthias; Rottner, Klemens; Brakebusch, Cord.

I: The Journal of Biological Chemistry, Bind 293, Nr. 24, 2018, s. 9358-9369.

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Harvard

Ge, J, Burnier, L, Adamopoulou, M, Kwa, MQ, Schaks, M, Rottner, K & Brakebusch, C 2018, 'RhoA, Rac1, and Cdc42 differentially regulate αSMA and collagen I expression in mesenchymal stem cells', The Journal of Biological Chemistry, bind 293, nr. 24, s. 9358-9369. https://doi.org/10.1074/jbc.RA117.001113

APA

Ge, J., Burnier, L., Adamopoulou, M., Kwa, M. Q., Schaks, M., Rottner, K., & Brakebusch, C. (2018). RhoA, Rac1, and Cdc42 differentially regulate αSMA and collagen I expression in mesenchymal stem cells. The Journal of Biological Chemistry, 293(24), 9358-9369. https://doi.org/10.1074/jbc.RA117.001113

Vancouver

Ge J, Burnier L, Adamopoulou M, Kwa MQ, Schaks M, Rottner K o.a. RhoA, Rac1, and Cdc42 differentially regulate αSMA and collagen I expression in mesenchymal stem cells. The Journal of Biological Chemistry. 2018;293(24):9358-9369. https://doi.org/10.1074/jbc.RA117.001113

Author

Ge, Jianfeng ; Burnier, Laurent ; Adamopoulou, Maria ; Kwa, Mei Qi ; Schaks, Matthias ; Rottner, Klemens ; Brakebusch, Cord. / RhoA, Rac1, and Cdc42 differentially regulate αSMA and collagen I expression in mesenchymal stem cells. I: The Journal of Biological Chemistry. 2018 ; Bind 293, Nr. 24. s. 9358-9369.

Bibtex

@article{7059c31cb0234bc08b0ed4c938d9db61,
title = "RhoA, Rac1, and Cdc42 differentially regulate αSMA and collagen I expression in mesenchymal stem cells",
abstract = "Mesenchymal stem cells (MSC) are suggested to be important progenitors of myofibroblasts in fibrosis. To understand the role of Rho GTPase signaling in TGFβ-induced myofibroblast differentiation of MSC, we generated a novel MSC line and its descendants lacking functional Rho GTPases and Rho GTPase signaling components. Unexpectedly, our data revealed that Rho GTPase signaling is required for TGFβ-induced expression of α-smooth muscle actin (αSMA) but not of collagen I α1 (col1a1). Whereas loss of RhoA and Cdc42 reduced αSMA expression, ablation of the Rac1 gene had the opposite effect. Although actin polymerization and MRTFa were crucial for TGFβ-induced αSMA expression, neither Arp2/3-dependent actin polymerization nor cofilin-dependent severing and depolymerization of F-actin were required. Instead, F-actin levels were dependent on cell contraction, and TGFβ-induced actin polymerization correlated with increased cell contraction mediated by RhoA and Cdc42. Finally, we observed impaired collagen I secretion in MSC lacking RhoA or Cdc42. These data give novel molecular insights into the role of Rho GTPases in TGFβ signaling and have implications for our understanding of MSC function in fibrosis.",
author = "Jianfeng Ge and Laurent Burnier and Maria Adamopoulou and Kwa, {Mei Qi} and Matthias Schaks and Klemens Rottner and Cord Brakebusch",
note = "{\textcopyright} 2018 by The American Society for Biochemistry and Molecular Biology, Inc.",
year = "2018",
doi = "10.1074/jbc.RA117.001113",
language = "English",
volume = "293",
pages = "9358--9369",
journal = "Journal of Biological Chemistry",
issn = "0021-9258",
publisher = "American Society for Biochemistry and Molecular Biology, Inc.",
number = "24",

}

RIS

TY - JOUR

T1 - RhoA, Rac1, and Cdc42 differentially regulate αSMA and collagen I expression in mesenchymal stem cells

AU - Ge, Jianfeng

AU - Burnier, Laurent

AU - Adamopoulou, Maria

AU - Kwa, Mei Qi

AU - Schaks, Matthias

AU - Rottner, Klemens

AU - Brakebusch, Cord

N1 - © 2018 by The American Society for Biochemistry and Molecular Biology, Inc.

PY - 2018

Y1 - 2018

N2 - Mesenchymal stem cells (MSC) are suggested to be important progenitors of myofibroblasts in fibrosis. To understand the role of Rho GTPase signaling in TGFβ-induced myofibroblast differentiation of MSC, we generated a novel MSC line and its descendants lacking functional Rho GTPases and Rho GTPase signaling components. Unexpectedly, our data revealed that Rho GTPase signaling is required for TGFβ-induced expression of α-smooth muscle actin (αSMA) but not of collagen I α1 (col1a1). Whereas loss of RhoA and Cdc42 reduced αSMA expression, ablation of the Rac1 gene had the opposite effect. Although actin polymerization and MRTFa were crucial for TGFβ-induced αSMA expression, neither Arp2/3-dependent actin polymerization nor cofilin-dependent severing and depolymerization of F-actin were required. Instead, F-actin levels were dependent on cell contraction, and TGFβ-induced actin polymerization correlated with increased cell contraction mediated by RhoA and Cdc42. Finally, we observed impaired collagen I secretion in MSC lacking RhoA or Cdc42. These data give novel molecular insights into the role of Rho GTPases in TGFβ signaling and have implications for our understanding of MSC function in fibrosis.

AB - Mesenchymal stem cells (MSC) are suggested to be important progenitors of myofibroblasts in fibrosis. To understand the role of Rho GTPase signaling in TGFβ-induced myofibroblast differentiation of MSC, we generated a novel MSC line and its descendants lacking functional Rho GTPases and Rho GTPase signaling components. Unexpectedly, our data revealed that Rho GTPase signaling is required for TGFβ-induced expression of α-smooth muscle actin (αSMA) but not of collagen I α1 (col1a1). Whereas loss of RhoA and Cdc42 reduced αSMA expression, ablation of the Rac1 gene had the opposite effect. Although actin polymerization and MRTFa were crucial for TGFβ-induced αSMA expression, neither Arp2/3-dependent actin polymerization nor cofilin-dependent severing and depolymerization of F-actin were required. Instead, F-actin levels were dependent on cell contraction, and TGFβ-induced actin polymerization correlated with increased cell contraction mediated by RhoA and Cdc42. Finally, we observed impaired collagen I secretion in MSC lacking RhoA or Cdc42. These data give novel molecular insights into the role of Rho GTPases in TGFβ signaling and have implications for our understanding of MSC function in fibrosis.

U2 - 10.1074/jbc.RA117.001113

DO - 10.1074/jbc.RA117.001113

M3 - Journal article

C2 - 29700112

VL - 293

SP - 9358

EP - 9369

JO - Journal of Biological Chemistry

JF - Journal of Biological Chemistry

SN - 0021-9258

IS - 24

ER -

ID: 199342309